Your browser doesn't support javascript.
loading
Gli1 Defines a Subset of Fibro-adipogenic Progenitors that Promote Skeletal Muscle Regeneration With Less Fat Accumulation.
Yao, Lutian; Tichy, Elisia D; Zhong, Leilei; Mohanty, Sarthak; Wang, Luqiang; Ai, Emily; Yang, Shuying; Mourkioti, Foteini; Qin, Ling.
Affiliation
  • Yao L; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
  • Tichy ED; Department of Orthopaedic Surgery, The First Hospital of China Medical University, Shenyang, China.
  • Zhong L; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
  • Mohanty S; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
  • Wang L; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
  • Ai E; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
  • Yang S; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
  • Mourkioti F; Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA.
  • Qin L; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
J Bone Miner Res ; 36(6): 1159-1173, 2021 06.
Article in En | MEDLINE | ID: mdl-33529374
ABSTRACT
Skeletal muscle has remarkable regenerative ability after injury. Mesenchymal fibro-adipogenic progenitors (FAPs) are necessary, active participants during this repair process, but the molecular signatures of these cells and their functional relevance remain largely unexplored. Here, using a lineage tracing mouse model (Gli1-CreER Tomato), we demonstrate that Gli1 marks a small subset of muscle-resident FAPs with elevated Hedgehog (Hh) signaling. Upon notexin muscle injury, these cells preferentially and rapidly expanded within FAPs. Ablation of Gli1+ cells using a DTA mouse model drastically reduced fibroblastic colony-forming unit (CFU-F) colonies generated by muscle cells and impaired muscle repair at 28 days. Pharmacologic manipulation revealed that Gli1+ FAPs rely on Hh signaling to increase the size of regenerating myofiber. Sorted Gli1+ FAPs displayed superior clonogenicity and reduced adipogenic differentiation ability in culture compared to sorted Gli1- FAPs. In a glycerol injury model, Gli1+ FAPs were less likely to give rise to muscle adipocytes compared to other FAPs. Further cell ablation and Hh activator/inhibitor treatments demonstrated their dual actions in enhancing myogenesis and reducing adipogenesis after injury. Examining single-cell RNA-sequencing dataset of FAPs from normal mice indicated that Gli1+ FAPs with increased Hh signaling provide trophic signals to myogenic cells while restrict their own adipogenic differentiation. Collectively, our findings identified a subpopulation of FAPs that play an essential role in skeletal muscle repair. © 2021 American Society for Bone and Mineral Research (ASBMR).
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Adipogenesis / Hedgehog Proteins Type of study: Prognostic_studies Limits: Animals Language: En Journal: J Bone Miner Res Journal subject: METABOLISMO / ORTOPEDIA Year: 2021 Document type: Article Affiliation country: Panama

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Adipogenesis / Hedgehog Proteins Type of study: Prognostic_studies Limits: Animals Language: En Journal: J Bone Miner Res Journal subject: METABOLISMO / ORTOPEDIA Year: 2021 Document type: Article Affiliation country: Panama